Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 183(5): 1234-1248.e25, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33113353

RESUMEN

Brain metastasis (br-met) develops in an immunologically unique br-met niche. Central nervous system-native myeloid cells (CNS-myeloids) and bone-marrow-derived myeloid cells (BMDMs) cooperatively regulate brain immunity. The phenotypic heterogeneity and specific roles of these myeloid subsets in shaping the br-met niche to regulate br-met outgrowth have not been fully revealed. Applying multimodal single-cell analyses, we elucidated a heterogeneous but spatially defined CNS-myeloid response during br-met outgrowth. We found Ccr2+ BMDMs minimally influenced br-met while CNS-myeloid promoted br-met outgrowth. Additionally, br-met-associated CNS-myeloid exhibited downregulation of Cx3cr1. Cx3cr1 knockout in CNS-myeloid increased br-met incidence, leading to an enriched interferon response signature and Cxcl10 upregulation. Significantly, neutralization of Cxcl10 reduced br-met, while rCxcl10 increased br-met and recruited VISTAHi PD-L1+ CNS-myeloid to br-met lesions. Inhibiting VISTA- and PD-L1-signaling relieved immune suppression and reduced br-met burden. Our results demonstrate that loss of Cx3cr1 in CNS-myeloid triggers a Cxcl10-mediated vicious cycle, cultivating a br-met-promoting, immune-suppressive niche.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/secundario , Quimiocina CXCL10/metabolismo , Terapia de Inmunosupresión , Células Mieloides/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Receptor 1 de Quimiocinas CX3C/metabolismo , Sistema Nervioso Central/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Interferones/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Neutralización , Fenotipo , Linfocitos T/inmunología , Transcriptoma/genética
2.
Curr Opin Hematol ; 30(4): 137-143, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37052294

RESUMEN

PURPOSE OF REVIEW: It is well described that tumor-directed aberrant myelopoiesis contributes to the generation of various myeloid populations with tumor-promoting properties. A growing number of recent studies have revealed the importance of the previously unappreciated roles of erythroid progenitor cells (EPCs) in the context of cancer, bringing the updated concept that altered erythropoiesis also facilitates tumor growth and progression. Better characterization of EPCs may provide attractive therapeutic opportunities. RECENT FINDINGS: EPCs represent a heterogeneous population. They exhibit crucial pro-tumor activities by secreting growth factors and modulating the immune response. Cancers induce potent EPC expansion and suppress their differentiation. Recent single-cell transcriptome and lineage tracking analyses have provided novel insight that tumor-induced EPCs are able to be transdifferentiated into immunosuppressive myeloid cells to limit T-cell function and immunotherapy. Therapeutic strategies targeting key factors of EPC-driven immunosuppression, reducing the amount of EPCs, and promoting EPC differentiation and maturation have been extensively investigated. SUMMARY: This review summarizes the current state of knowledge as to the fascinating biology of EPCs, highlights mechanisms by which they exert the tumor promoting activities, as well as the perspectives on future directions and strategies to target these cells for potential therapeutic benefit.


Asunto(s)
Células Precursoras Eritroides , Neoplasias , Humanos , Diferenciación Celular , Neoplasias/terapia
3.
Int J Mol Sci ; 23(17)2022 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-36077309

RESUMEN

BACKGROUND: Mechanically gated PIEZO channels lead to an influx of cations, activation of additional Ca2+ channels, and cell depolarization. This study aimed to investigate PIEZO2's role in breast cancer. METHODS: The clinical relevance of PIEZO2 expression in breast cancer patient was analyzed in a publicly available dataset. Utilizing PIEZO2 overexpressed breast cancer cells, and in vitro and in vivo experiments were conducted. RESULTS: High expression of PIEZO2 was correlated with a worse survival in triple-negative breast cancer (TNBC) but not in other subtypes. Increased PEIZO2 channel function was confirmed in PIEZO2 overexpressed cells after mechanical stimulation. PIEZO2 overexpressed cells showed increased motility and invasive phenotypes as well as higher expression of SNAIL and Vimentin and lower expression of E-cadherin in TNBC cells. Correspondingly, high expression of PIEZO2 was correlated with the increased expression of epithelial-mesenchymal transition (EMT)-related genes in a TNBC patient. Activated Akt signaling was observed in PIEZO2 overexpressed TNBC cells. PIEZO2 overexpressed MDA-MB-231 cells formed a significantly higher number of lung metastases after orthotopic implantation. CONCLUSION: PIEZO2 activation led to enhanced SNAIL stabilization through Akt activation. It enhanced Vimentin and repressed E-cadherin transcription, resulting in increased metastatic potential and poor clinical outcomes in TNBC patients.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Humanos , Canales Iónicos/genética , Fenotipo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Vimentina/genética , Vimentina/metabolismo
4.
Small ; 11(21): 2511-7, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-25688868

RESUMEN

Cobalt sulfide (CoS2) is considered one of the most promising alternative anode materials for high-performance lithium-ion batteries (LIBs) by virtue of its remarkable electrical conductivity, high theoretical capacity, and low cost. However, it suffers from a poor cycling stability and low rate capability because of its volume expansion and dissolution of the polysulfide intermediates in the organic electrolytes during the battery charge/discharge process. In this study, a novel porous carbon/CoS2 composite is prepared by using nano metal-organic framework (MOF) templates for high-preformance LIBs. The as-made ultrasmall CoS2 (15 nm) nanoparticles in N-rich carbon exhibit promising lithium storage properties with negligible loss of capacity at high charge/discharge rate. At a current density of 100 mA g(-1), a capacity of 560 mA h g(-1) is maintained after 50 cycles. Even at a current density as high as 2500 mA g(-1), a reversible capacity of 410 mA h g(-1) is obtained. The excellent and highly stable battery performance should be attributed to the synergism of the ultrasmall CoS2 particles and the thin N-rich porous carbon shells derieved from nanosized MOF precusors.

5.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 43(4): 427-33, 2014 07.
Artículo en Zh | MEDLINE | ID: mdl-25187457

RESUMEN

OBJECTIVE: To generate two genetically engineered mouse models of ErbB2/Neu positive-PTEN deficient breast cancer and to compare their biological properties. METHODS: The genetically engineered mice previously developed with mouse mammary tumor virus (MMTV) promoter driven expression of activated ErbB2/Neu and recombinant Cre (FVB/N-MMTV-NIC) were interbred with Flox-PTEN mice; and FVB/N-ErbB2KI mice, harboring endogenous promoter driven activated ErbB2/Neu expression, FVB/N-MMTV-Cre mice and the flox-PTEN mice were interbred. Neu, Cre and PTEN genes were amplified by PCR for genotyping of the offsprings. ErbB2/Neu and PTEN expression in mammary tumors were detected by immunohistochemistry. Tumor formation time, tumor number, histopathology and lung metastasis were compared between two models, Ki-67 expression was detected by immunohistochemistry, and TUNEL staining of tumor tissues was performed. RESULTS: Two genetically engineered mouse models of ErbB2/Neu positive-PTEN homozygous deficient breast cancer were generated. The models were confirmed by genotyping and immunohistochemistry. One model with exogenous MMTV promoter driven expression of activated ErbB2/Neu and Cre coupling PTEN disruption was designated as NIC/PTEN(-/-) mice, and the other with MMTV-Cre induced endogenous promoter driven expression of activated ErbB2/Neu with PTEN disruption was designated as ErbB2KI/PTEN(-/-) mice. The tumor formation time in NIC/PTEN(-/-) mice was significantly shorter than that of ErbB2KI/PTEN(-/-) mice (30 vs 368 d, P<0.01); the number of tumor and incidence of lung metastasis was also significantly higher in NIC/PTEN(-/-) mice (10 vs 1-2 and 75.0% vs 37.5%, respectively, Ps<0.01). The Two models displayed distinct histopathological morphology. NIC/PTEN(-/-) tumor showed more Ki-67 positive cells than ErbB2KI/PTEN(-/-) tumor did (86.9%±2.8% vs 37.4%±7.2%, P<0.01), while the amount of cell apoptosis in tumors was not significantly different between two models. CONCLUSION: Two genetically engineered mouse models of ErbB2/Neu positive-PTEN homozygous deficient breast cancer with different phenotypes have been successfully generated, which may provide useful resource for further investigation of the initiation and progression of HER2/ErbB2 breast cancer, as well as for the development of novel prevention and treatment regimens of this malignance.


Asunto(s)
Neoplasias de la Mama/genética , Modelos Animales de Enfermedad , Neoplasias Mamarias Animales , Fosfohidrolasa PTEN/genética , Receptor ErbB-2/genética , Animales , Femenino , Eliminación de Gen , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Transgénicos
6.
Cancers (Basel) ; 16(4)2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38398074

RESUMEN

PIEZO1 plays a crucial role in the human body as a mechanosensory ion channel. It has been demonstrated that PIEZO1 is important in tissue development and regulating many essential physiological processes. Studies have suggested that the PIEZO1 ion channel plays a role in invasion and progression in cancer; elevated levels of PIEZO1 have been correlated with increased migration in breast cancer cells, chemo-resistance and invasion in gastric cancer cells, and increased invasion of osteosarcoma cells. In addition, high PIEZO1 expression levels were correlated with a worse prognosis in glioma patients. On the other hand, studies in lung cancer have attributed high PIEZO1 levels to better patient outcomes. However, the clinical impact of PIEZO1 in breast cancer is not well characterized. Therefore, our goal was to determine the clinical relevance of PIEZO1 in breast cancer. An analysis of breast cancer data from The Cancer Genome Atlas (TCGA) was conducted to investigate PIEZO1 expression levels and correlation to survival, followed by validation in an independent dataset, GSE3494. We also performed gene set enrichment analysis (GSEA) and pathway enrichment analysis. We also analyzed the immune cell composition in breast tumors from TCGA through a CIBERSORT algorithm. Our results demonstrated that the PIEZO1 expression levels are higher in hormone-receptor (HR)-negative than in HR-positive cohorts. High PIEZO1 expression is correlated with a significant decrease in survival in HR-negative cohorts, especially in triple-negative breast cancer (TNBC), suggesting that PIEZO1 could be utilized as a prognostic biomarker in HR-negative breast cancer. GSEA showed that various signaling pathways associated with more invasive phenotypes and resistance to treatments, including epithelial-mesenchymal transition (EMT), hypoxia, and multiple signaling pathways, are enriched in high-PIEZO1 HR-negative tumors. Our results also demonstrated a decrease in CD8+ and CD4+ T cell infiltration in high-PIEZO1 HR-negative tumors. Further investigations are necessary to elucidate the mechanistic roles of PIEZO1 in HR-negative breast cancer.

7.
Natl Sci Rev ; 10(3): nwac257, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36879845

RESUMEN

This study, via combined analysis of geophysical and geochemical data, reveals a lithospheric architecture characterized by crust-mantle decoupling and vertical heat-flow conduits that control orogenic gold mineralization in the Ailaoshan gold belt on the southeastern margin of Tibet. The mantle seismic tomography indicates that the crust-mantle decoupled deformation, defined from previous seismic anisotropy analysis, was formed by upwelling and lateral flow of the asthenosphere, driven by deep subduction of the Indian continent. Our magnetotelluric and seismic images show both a vertical conductor across the Moho and high Vp/Vs anomalies both in the uppermost mantle and lowest crust, suggesting that crust-mantle decoupling promotes ponding of mantle-derived basic melts at the base of the crust via a heat-flow conduit. Noble gas isotope and halogen ratios of gold-related ore minerals indicate a mantle source of ore fluid. A rapid decrease in Cl/F ratios of lamprophyres under conditions of 1.2 GPa and 1050°C suggests that the ore fluid was derived from degassing of the basic melts. Similar lithospheric architecture is recognized in other orogenic gold provinces, implying analogous formational controls.

8.
Nat Commun ; 14(1): 2109, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37055410

RESUMEN

Chemotherapy prior to immune checkpoint blockade (ICB) treatment appears to improve ICB efficacy but resistance to ICB remains a clinical challenge and is attributed to highly plastic myeloid cells associating with the tumor immune microenvironment (TIME). Here we show by CITE-seq single-cell transcriptomic and trajectory analyses that neoadjuvant low-dose metronomic chemotherapy (MCT) leads to a characteristic co-evolution of divergent myeloid cell subsets in female triple-negative breast cancer (TNBC). Specifically, we identify that the proportion of CXCL16 + myeloid cells increase and a high STAT1 regulon activity distinguishes Programmed Death Ligand 1 (PD-L1) expressing immature myeloid cells. Chemical inhibition of STAT1 signaling in MCT-primed breast cancer sensitizes TNBC to ICB treatment, which underscores the STAT1's role in modulating TIME. In summary, we leverage single-cell analyses to dissect the cellular dynamics in the tumor microenvironment (TME) following neoadjuvant chemotherapy and provide a pre-clinical rationale for modulating STAT1 in combination with anti-PD-1 for TNBC patients.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Radioinmunoterapia , Células Mieloides , Quimiocina CXCL16 , Microambiente Tumoral , Factor de Transcripción STAT1/genética
9.
Cell Metab ; 35(10): 1688-1703.e10, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37793345

RESUMEN

Metastasis causes breast cancer-related mortality. Tumor-infiltrating neutrophils (TINs) inflict immunosuppression and promote metastasis. Therapeutic debilitation of TINs may enhance immunotherapy, yet it remains a challenge to identify therapeutic targets highly expressed and functionally essential in TINs but under-expressed in extra-tumoral neutrophils. Here, using single-cell RNA sequencing to compare TINs and circulating neutrophils in murine mammary tumor models, we identified aconitate decarboxylase 1 (Acod1) as the most upregulated metabolic enzyme in mouse TINs and validated high Acod1 expression in human TINs. Activated through the GM-CSF-JAK/STAT5-C/EBPß pathway, Acod1 produces itaconate, which mediates Nrf2-dependent defense against ferroptosis and upholds the persistence of TINs. Acod1 ablation abates TIN infiltration, constrains metastasis (but not primary tumors), bolsters antitumor T cell immunity, and boosts the efficacy of immune checkpoint blockade. Our findings reveal how TINs escape from ferroptosis through the Acod1-dependent immunometabolism switch and establish Acod1 as a target to offset immunosuppression and improve immunotherapy against metastasis.


Asunto(s)
Neoplasias de la Mama , Carboxiliasas , Ferroptosis , Humanos , Ratones , Animales , Femenino , Neoplasias de la Mama/metabolismo , Neutrófilos , Carboxiliasas/metabolismo , Melanoma Cutáneo Maligno
10.
J Nanosci Nanotechnol ; 12(5): 3812-20, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22852311

RESUMEN

A modified solid-state method was used to prepare LiFePO4. With the aid of deionized water, a mixture containing Fe2O3, NH4H2PO4 (or (NH4)2HPO4), LiOH, glucose and oxalic acid was prepared into fluffy powders, which were heated in a carbon-coated crucible at 700 degrees C for 3 hours to synthesize LiFePO4 without any inert gas flow. For the first time, the roles of NH4H2PO4 and (NH4)2HPO4 on the preparation of LiFePO4 were systematically investigated. The obtained samples were characterized by X-ray diffraction (XRD), scanning electron microscope (SEM) and energy dispersive spectroscopy (EDS), revealing that the crystallinity of the LiFePO4 sample prepared from NH4H2PO4 is superior to that prepared from (NH4)2HPO4 and the particle size of the sample prepared from NH4H2PO4 is smaller than that prepared from (NH4)2HPO4. The specific capacity, cycle property and rate capabilities were also compared between the as-prepared LiFePO4 samples. A better electrochemical performance was observed in the sample prepared from NH4H2PO4.

11.
IEEE Trans Biomed Eng ; 68(11): 3317-3326, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33793396

RESUMEN

GOAL: Typical SRUS images are reconstructed by localizing ultrasound microbubbles (MBs) injected in a vessel using normalized 2-dimensional cross-correlation (2DCC) between MBs signals and the point spread function of the system. However, current techniques require isolated MBs in a confined area due to inaccurate localization of densely populated MBs. To overcome this limitation, we developed the ℓ1-homotopy based compressed sensing (L1H-CS) based SRUS imaging technique which localizes densely populated MBs to visualize microvasculature in vivo. METHODS: To evaluate the performance of L1H-CS, we compared the performance of 2DCC, interior-point method based compressed sensing (CVX-CS), and L1H-CS algorithms. Localization efficiency was compared using axially and laterally aligned point targets (PTs) with known distances and randomly distributed PTs generated by simulation. We developed post-processing techniques including clutter reduction, noise equalization, motion compensation, and spatiotemporal noise filtering for in vivo imaging. We then validated the capabilities of L1H-CS based SRUS imaging technique with high-density MBs in a mouse tumor model, kidney, and zebrafish dorsal trunk, and brain. RESULTS: Compared to 2DCC and CVX-CS algorithms, L1H-CS achieved faster data acquisition time and considerable improvement in SRUS image quality. CONCLUSIONS AND SIGNIFICANCE: These results demonstrate that the L1H-CS based SRUS imaging technique has the potential to examine microvasculature with reduced acquisition and reconstruction time to acquire enhanced SRUS image quality, which may be necessary to translate it into clinics.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Pez Cebra , Algoritmos , Animales , Imagen por Resonancia Magnética , Ratones , Movimiento (Física) , Ultrasonografía
12.
STAR Protoc ; 2(2): 100537, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34036283

RESUMEN

High dimensional compositional and transcriptional profiling of heterogeneous brain-infiltrating leukocytes can lead to novel biological and therapeutic discoveries. High-quality single-cell leukocyte preparations are a prerequisite for optimal single cell profiling. Here, we describe a protocol for epitope and RNA-preserving dissociation of adult mouse brains and subsequent leukocyte purification and staining, which is adaptable to homeostatic and pathogenic brains. Leukocyte preparation following this protocol permits exquisite single-cell surface protein and RNA profiling in applications including CyTOF and CITE-seq. For complete details on the use and execution of this protocol, please refer to Guldner et al. (2020) and Golomb et al. (2020).


Asunto(s)
Encéfalo/patología , Separación Celular/métodos , Epítopos/genética , Leucocitos , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Animales , Células Cultivadas , Femenino , Leucocitos/citología , Leucocitos/metabolismo , Masculino , Ratones
13.
Front Pharmacol ; 12: 697704, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34421596

RESUMEN

Objective: To explore the molecular mechanism of Scutellaria baicalensis Georgi in treating gastric cancer by network pharmacological analysis and molecular docking. Methods: Taking Scutellaria baicalensis Georgi as the object, the active components and corresponding potential drug targets in Scutellaria baicalensis Georgi were obtained from the database of TCM Pharmacological System Analysis Platform (TCMSP). GeneCards/OMIM/DrugBank and other databases were used to collect gastric cancer-related genes, and the obtained genes were intersected with drug targets to obtain the target genes of Scutellaria baicalensis Georgi on gastric cancer. Furthermore, the interaction network of Scutellaria baicalensis Georgi-active ingredients-target-gastric cancer-related genes was constructed. Protein-protein interaction analysis and gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed on target genes. The PubChem website was used to screen the compounds corresponding to the target genes, and the target protein and 3D structure pdb format files were obtained from the PDB database. Finally, the molecular docking calculation was performed by the AutoDock Vina program. The in vivo cell experiments on the effect of Scutellaria baicalensis on proliferation and migration of gastric cancer cells were used to determine the therapeutic effect of Scutellaria baicalensis on gastric cancer, and the two genes ESR1 and FOS are the key targets of Scutellaria baicalensis on gastric cancer. Results: A total of 10 gastric cancer-related target genes were screened out, and Scutellaria baicalensis Georgi contained 10 active compounds targeting 10 gene sites. There are 30 effective compounds in Scutellaria baicalensis Georgi targeted to treat gastric cancer, and there are 91 corresponding targeting gene sites, involving a total of 10 pathways. The results of molecular docking show that ESR1, FOS, and Scutellaria baicalensis Georgi have good binding free energy and docking fraction. The docking fraction of FOS is -4.200 and the binding free energy is -27.893 kcal/mol. The docking fraction of ESR1 is -5.833 and the binding free energy is -30.001 kcal/mol. The effect of Scutellaria baicalensis Georgi on gastric cancer was verified by in vitro cell experiments and Western blotting. Conclusion: Scutellaria baicalensis Georgi can target and regulate multiple signal pathways by acting on ESR1 and FOS gene loci, thus having a potential therapeutic effect on gastric cancer.

14.
J Clin Invest ; 131(4)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33320840

RESUMEN

Rapidly proliferating tumor and immune cells need metabolic programs that support energy and biomass production. The amino acid glutamine is consumed by effector T cells and glutamine-addicted triple-negative breast cancer (TNBC) cells, suggesting that a metabolic competition for glutamine may exist within the tumor microenvironment, potentially serving as a therapeutic intervention strategy. Here, we report that there is an inverse correlation between glutamine metabolic genes and markers of T cell-mediated cytotoxicity in human basal-like breast cancer (BLBC) patient data sets, with increased glutamine metabolism and decreased T cell cytotoxicity associated with poor survival. We found that tumor cell-specific loss of glutaminase (GLS), a key enzyme for glutamine metabolism, improved antitumor T cell activation in both a spontaneous mouse TNBC model and orthotopic grafts. The glutamine transporter inhibitor V-9302 selectively blocked glutamine uptake by TNBC cells but not CD8+ T cells, driving synthesis of glutathione, a major cellular antioxidant, to improve CD8+ T cell effector function. We propose a "glutamine steal" scenario, in which cancer cells deprive tumor-infiltrating lymphocytes of needed glutamine, thus impairing antitumor immune responses. Therefore, tumor-selective targeting of glutamine metabolism may be a promising therapeutic strategy in TNBC.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos T CD8-positivos/inmunología , Proteínas Portadoras/antagonistas & inhibidores , Glutamina/inmunología , Inmunidad Celular , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias de la Mama Triple Negativas/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Femenino , Glutamina/metabolismo , Xenoinjertos , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
15.
Cancer Lett ; 503: 163-173, 2021 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-33524500

RESUMEN

The majority of women with ovarian cancer are diagnosed with metastatic disease, therefore elucidating molecular events that contribute to successful metastatic dissemination may identify additional targets for therapeutic intervention and thereby positively impact survival. Using two human high grade serous ovarian cancer cell lines with inactive TP53 and multiple rounds of serial in vivo passaging, we generated sublines with significantly accelerated intra-peritoneal (IP) growth. Comparative analysis of the parental and IP sublines identified a common panel of differentially expressed genes. The most highly differentially expressed gene, upregulated by 60-65-fold in IP-selected sublines, was the type I transmembrane protein AMIGO2. As the role of AMIGO2 in ovarian cancer metastasis remains unexplored, CRISPR/Cas9 was used to reduce AMIGO2 expression, followed by in vitro and in vivo functional analyses. Knockdown of AMIGO2 modified the sphere-forming potential of ovarian cancer cells, reduced adhesion and invasion in vitro, and significantly attenuated IP metastasis. These data highlight AMIGO2 as a new target for a novel anti-metastatic therapeutic approach aimed at blocking cohesion, survival, and adhesion of metastatic tumorspheres.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Ováricas/patología , Neoplasias Peritoneales/patología , Neoplasias Peritoneales/secundario , Regulación hacia Arriba , Animales , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Mutación , Trasplante de Neoplasias , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/metabolismo , Proteína p53 Supresora de Tumor/genética
16.
Cell Rep ; 33(9): 108438, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33264626

RESUMEN

Phenotypic and functional plasticity of brain immune cells contribute to brain tissue homeostasis and disease. Immune cell plasticity is profoundly influenced by tissue microenvironment cues and systemic factors. Aging and gut microbiota dysbiosis that reshape brain immune cell plasticity and homeostasis has not been fully delineated. Using Cellular Indexing of Transcriptomes and Epitopes by sequencing (CITE-seq), we analyze compositional and transcriptional changes of the brain immune landscape in response to aging and gut dysbiosis. Discordance between canonical surface-marker-defined immune cell types and their transcriptomes suggest transcriptional plasticity among immune cells. Ly6C+ monocytes predominate a pro-inflammatory signature in the aged brain, while innate lymphoid cells (ILCs) shift toward an ILC2-like profile. Aging increases ILC-like cells expressing a T memory stemness (Tscm) signature, which is reduced through antibiotics-induced gut dysbiosis. Systemic changes due to aging and gut dysbiosis increase propensity for neuroinflammation, providing insights into gut dysbiosis in age-related neurological diseases.


Asunto(s)
Encéfalo/inmunología , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Innata/inmunología , Análisis de la Célula Individual/métodos , Animales , Humanos
17.
JCI Insight ; 5(15)2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32759497

RESUMEN

A tumor blood vessel is a key regulator of tissue perfusion, immune cell trafficking, cancer metastasis, and therapeutic responsiveness. mTORC1 is a signaling node downstream of multiple angiogenic factors in the endothelium. However, mTORC1 inhibitors have limited efficacy in most solid tumors, in part due to inhibition of immune function at high doses used in oncology patients and compensatory PI3K signaling triggered by mTORC1 inhibition in tumor cells. Here we show that low-dose RAD001/everolimus, an mTORC1 inhibitor, selectively targets mTORC1 signaling in endothelial cells (ECs) without affecting tumor cells or immune cells, resulting in tumor vessel normalization and increased antitumor immunity. Notably, this phenotype was recapitulated upon targeted inducible gene ablation of the mTORC1 component Raptor in tumor ECs (RaptorECKO). Tumors grown in RaptorECKO mice displayed a robust increase in tumor-infiltrating lymphocytes due to GM-CSF-mediated activation of CD103+ dendritic cells and displayed decreased tumor growth and metastasis. GM-CSF neutralization restored tumor growth and metastasis, as did T cell depletion. Importantly, analyses of human tumor data sets support our animal studies. Collectively, these findings demonstrate that endothelial mTORC1 is an actionable target for tumor vessel normalization, which could be leveraged to enhance antitumor immune therapies.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Everolimus/farmacología , Linfocitos Infiltrantes de Tumor/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
18.
J Nanosci Nanotechnol ; 9(4): 2316-23, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19437970

RESUMEN

Carbon nanofibers (CNFs) with different microstructures, including platelet-CNFs (PCNFs), fish-bone-CNFs, and tube-CNFs, were synthesized, characterized and evaluated toward methanol oxidation reaction (MOR). The CNFs studied here showed several structures in which various stacked morphologies as well as the ordering of their size and graphite layers can be well controlled. Platinum nanoparticles have been electrodeposited on CNFs surfaces, and their electrocatalytic activities toward MOR have been studied using cyclic voltammetry, chronoamperometry, and linear sweep voltammograms. Morphologies, textural properties, and the crystalline structure of the CNFs supports and catalysts have been characterized with transmission electron microscopy and scanning electron microscopy. The comparative tests conclude that Pt/PCNFs have the best electrocatalytic performance and good stability at room temperature. The high electrocatalytic activity and stability can be attributed to the specific microstructure of PCNFs, which have large numbers of edge-active carbon atoms on the surface of the CNFs as well as synergistic effects between CNFs and platinum nanoparticles. The results suggest that PCNFs are excellent potential candidates as catalyst supports in direct methanol fuel cells.

19.
Nat Commun ; 10(1): 3817, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31444334

RESUMEN

Acquired resistance to targeted cancer therapy is a significant clinical challenge. In parallel with clinical trials combining CDK4/6 inhibitors to treat HER2+ breast cancer, we sought to prospectively model tumor evolution in response to this regimen in vivo and identify a clinically actionable strategy to combat drug resistance. Despite a promising initial response, acquired resistance emerges rapidly to the combination of anti-HER2/neu antibody and CDK4/6 inhibitor Palbociclib. Using high-throughput single-cell profiling over the course of treatments, we reveal a distinct immunosuppressive immature myeloid cell (IMC) population to infiltrate the resistant tumors. Guided by single-cell transcriptome analysis, we demonstrate that combination of IMC-targeting tyrosine kinase inhibitor cabozantinib and immune checkpoint blockade enhances anti-tumor immunity, and overcomes the resistance. Furthermore, sequential combinatorial immunotherapy enables a sustained control of the fast-evolving CDK4/6 inhibitor-resistant tumors. Our study demonstrates a translational framework for treating rapidly evolving tumors through preclinical modeling and single-cell analyses.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Modelos Biológicos , Células Progenitoras Mieloides/efectos de los fármacos , Células Progenitoras Mieloides/inmunología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Análisis de la Célula Individual , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Transpl Immunol ; 19(2): 145-51, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18503890

RESUMEN

BACKGROUND AND PURPOSE: Since the Fas/Fas Ligand (FasL) interaction has been recognized as an apoptotic pathway, it eliminates the activated T cells and promotes the survival of grafts. In this study, the effect of FasL transfection of pig chondrocytes on allogeneic transplantation was examined in vitro and in vivo. METHODS: Chondrocytes were isolated from articular and aural cartilages of anesthetized Guizhou Xiang (Gz) pig. The cells were transfected with G418 selected virus, packed from PA317 cells with a constructed plasmid using pig FasL (pGCEN-FasL). The apoptotic effect of FasL transfection was examined on Jurkat cells and activated recipient Gz T cells. The FasL expression was assessed by Western blot and flow cytometry. FasL+chondrocytes-Pluronic F-127 complex was injected into the right abdomen of recipient Gz pig. Histology and morphology of the engineered tissue were examined after 2 and 5 weeks of transplantation. RESULTS: The FasL expression was confirmed in pGCEN-FasL transfected chondrocytes. The expression of FasL of chondrocytes from Gz pig was analyzed by FACS. The apoptosis of Jurkat cells and activated recipient Gz T cells was increased by co-culture with FasL(+) chondrocytes (53.41% and 30.38% (E/T=10:1), in contrast of 32.27% and 13.16% with the control chondrocytes, respectively, P<0.01). FasL(+) chondrocytes-Pluronic F-127 implant expressed FasL and Type II collagen at the 5th week and survived until the 8th week. INTERPRETATION: The result indicates that the expression of FasL by chondrocytes is capable of inducing apoptosis of activated T cells. This suggests a potential role for allogeneic transplantation with chondrocytes.


Asunto(s)
Condrocitos/inmunología , Condrocitos/trasplante , Colágeno Tipo II/metabolismo , Proteína Ligando Fas/metabolismo , Linfocitos T/inmunología , Animales , Apoptosis , Condrocitos/citología , Condrocitos/metabolismo , Colágeno Tipo II/inmunología , Proteína Ligando Fas/genética , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Humanos , Células Jurkat , Activación de Linfocitos , Masculino , Porcinos , Porcinos Enanos , Linfocitos T/citología , Linfocitos T/metabolismo , Ingeniería de Tejidos , Transfección , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA